Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep Methods ; 3(1): 100372, 2023 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-36814844

RESUMO

The development of protease-activatable drugs and diagnostics requires identifying substrates specific to individual proteases. However, this process becomes increasingly difficult as the number of target proteases increases because most substrates are promiscuously cleaved by multiple proteases. We introduce a method-substrate libraries for compressed sensing of enzymes (SLICE)-for selecting libraries of promiscuous substrates that classify protease mixtures (1) without deconvolution of compressed signals and (2) without highly specific substrates. SLICE ranks substrate libraries using a compression score (C), which quantifies substrate orthogonality and protease coverage. This metric is predictive of classification accuracy across 140 in silico (Pearson r = 0.71) and 55 in vitro libraries (r = 0.55). Using SLICE, we select a two-substrate library to classify 28 samples containing 11 enzymes in plasma (area under the receiver operating characteristic curve [AUROC] = 0.93). We envision that SLICE will enable the selection of libraries that capture information from hundreds of enzymes using fewer substrates for applications like activity-based sensors for imaging and diagnostics.


Assuntos
Endopeptidases , Peptídeo Hidrolases , Especificidade por Substrato , Peptídeo Hidrolases/metabolismo
2.
Nat Biomed Eng ; 6(3): 310-324, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35241815

RESUMO

Immune checkpoint blockade (ICB) therapy does not benefit the majority of treated patients, and those who respond to the therapy can become resistant to it. Here we report the design and performance of systemically administered protease activity sensors conjugated to anti-programmed cell death protein 1 (αPD1) antibodies for the monitoring of antitumour responses to ICB therapy. The sensors consist of a library of mass-barcoded protease substrates that, when cleaved by tumour proteases and immune proteases, are released into urine, where they can be detected by mass spectrometry. By using syngeneic mouse models of colorectal cancer, we show that random forest classifiers trained on mass spectrometry signatures from a library of αPD1-conjugated mass-barcoded activity sensors for differentially expressed tumour proteases and immune proteases can be used to detect early antitumour responses and discriminate resistance to ICB therapy driven by loss-of-function mutations in either the B2m or Jak1 genes. Biomarkers of protease activity may facilitate the assessment of early responses to ICB therapy and the classification of refractory tumours based on resistance mechanisms.


Assuntos
Imunoconjugados , Neoplasias , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Peptídeo Hidrolases , Urinálise
3.
Sci Adv ; 8(8): eabm7950, 2022 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-35196075

RESUMO

Simultaneous delivery of mRNA to multiple populations of antigen (Ag)-specific CD8+ T cells is challenging given the diversity of peptide epitopes and polymorphism of class I major histocompatibility complexes (MHCI). We developed Ag-presenting nanoparticles (APNs) for mRNA delivery using pMHCI molecules that were refolded with photocleavable peptides to allow rapid ligand exchange by UV light and site-specifically conjugated with a lipid tail for postinsertion into preformed mRNA lipid nanoparticles. Across different TCR transgenic mouse models (P14, OT-1, and Pmel), UV-exchanged APNs bound and transfected their cognate Ag-specific CD8+ T cells equivalent to APNs produced using conventionally refolded pMHCI molecules. In mice infected with PR8 influenza, multiplexed delivery of UV-exchanged APNs against three immunodominant epitopes led to ~50% transfection of a VHH mRNA reporter in cognate Ag-specific CD8+ T cells. Our data show that UV-mediated peptide exchange can be used to rapidly produce APNs for mRNA delivery to multiple populations of Ag-specific T cells in vivo.

4.
Adv Sci (Weinh) ; 8(19): e2102043, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34363349

RESUMO

The deficiency of antigen-specific T cells and the induction of various treatment-induced immunosuppressions still limits the clinical benefit of cancer immunotherapy. Although the chemo-immunotherapy adjuvanted with Toll-like receptor 7/8 agonist (TLR 7/8a) induces immunogenic cell death (ICD) and in situ vaccination effect, indoleamine 2,3-dioxygenase (IDO) is also significantly increased in the tumor microenvironment (TME) and tumor-draining lymph node (TDLN), which offsets the activated antitumor immunity. To address the treatment-induced immunosuppression, an assemblable immune modulating suspension (AIMS) containing ICD inducer (paclitaxel) and supra-adjuvant (immune booster; R848 as a TLR 7/8a, immunosuppression reliever; epacadostat as an IDO inhibitor) is suggested and shows that it increases cytotoxic T lymphocytes and relieves the IDO-related immunosuppression (TGF-ß, IL-10, myeloid-derived suppressor cells, and regulatory T cells) in both TME and TDLN, by the formation of in situ depot in tumor bed as well as by the efficient migration into TDLN. Local administration of AIMS increases T cell infiltration in both local and distant tumors and significantly inhibits the metastasis of tumors to the lung. Reverting treatment-induced secondary immunosuppression and reshaping "cold tumor" into "hot tumor" by AIMS also increases the response rate of immune checkpoint blockade therapy, which promises a new nanotheranostic strategy in cancer immunotherapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Terapia de Imunossupressão/métodos , Imunoterapia/métodos , Nanomedicina/métodos , Animais , Modelos Animais de Doenças , Imunoterapia/efeitos adversos
5.
Nat Biomed Eng ; 5(11): 1348-1359, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34385695

RESUMO

Treating solid malignancies with chimeric antigen receptor (CAR) T cells typically results in poor responses. Immunomodulatory biologics delivered systemically can augment the cells' activity, but off-target toxicity narrows the therapeutic window. Here we show that the activity of intratumoural CAR T cells can be controlled photothermally via synthetic gene switches that trigger the expression of transgenes in response to mild temperature elevations (to 40-42 °C). In vitro, heating engineered primary human T cells for 15-30 min led to over 60-fold-higher expression of a reporter transgene without affecting the cells' proliferation, migration and cytotoxicity. In mice, CAR T cells photothermally heated via gold nanorods produced a transgene only within the tumours. In mouse models of adoptive transfer, the systemic delivery of CAR T cells followed by intratumoural production, under photothermal control, of an interleukin-15 superagonist or a bispecific T cell engager bearing an NKG2D receptor redirecting T cells against NKG2D ligands enhanced antitumour activity and mitigated antigen escape. Localized photothermal control of the activity of engineered T cells may enhance their safety and efficacy.


Assuntos
Receptores de Antígenos Quiméricos , Animais , Deriva e Deslocamento Antigênicos , Linhagem Celular Tumoral , Fatores Imunológicos , Imunoterapia Adotiva , Camundongos , Receptores de Antígenos Quiméricos/genética , Linfócitos T
6.
Adv Mater ; 31(42): e1903242, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31490604

RESUMO

Current cancer immunotherapy based on immune checkpoint blockade (ICB) still suffers from low response rate and systemic toxicity. To overcome the limitation, a novel therapeutic platform that can revert nonimmunogenic tumors into immunogenic phenotype is highly required. Herein, a designer scaffold loaded with both immune nanoconverters encapsulated with resiquimod (iNCVs (R848)) and doxorubicin, which provides the polarization of immunosuppressive tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) into tumoricidal antigen-presenting cells (APCs), rather than depleting them, as well as in situ vaccination that can be generated in vivo without the need to previously analyze and sequence tumor antigens to favor neoantigen-specific T cell responses is suggested. Local and sustained release of iNCVs (R848) and doxorubicin from the designer scaffold not only reduces the frequency of immunosuppressive cells in tumors but also increases systemic antitumor immune response, while minimizing systemic toxicity. Reshaping the tumor microenivronment (TME) using the designer-scaffold-induced synergistic antitumor immunity with ICB effects and long-term central and effector memory T cell responses, results in the prevention of postsurgical tumor recurrence and metastasis. The spatiotemporal modulation of TMEs through designer scaffolds is expected to be a strategy to overcome the limitations and improve the therapeutic efficacy of current immunotherapies with minimized systemic toxicity.


Assuntos
Tolerância Imunológica/imunologia , Imunoterapia/métodos , Nanomedicina/métodos , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Doxorrubicina/química , Doxorrubicina/farmacologia , Imidazóis/química , Camundongos
7.
Nat Commun ; 10(1): 3745, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31431623

RESUMO

The low response rate of current cancer immunotherapy suggests the presence of few antigen-specific T cells and a high number of immunosuppressive factors in tumor microenvironment (TME). Here, we develop a syringeable immunomodulatory multidomain nanogel (iGel) that overcomes the limitation by reprogramming of the pro-tumoral TME to antitumoral immune niches. Local and extended release of immunomodulatory drugs from iGel deplete immunosuppressive cells, while inducing immunogenic cell death and increased immunogenicity. When iGel is applied as a local postsurgical treatment, both systemic antitumor immunity and a memory T cell response are generated, and the recurrence and metastasis of tumors to lungs and other organs are significantly inhibited. Reshaping of the TME using iGel also reverts non-responding groups to checkpoint blockade therapies into responding groups. The iGel is expected as an immunotherapeutic platform that can reshape immunosuppressive TMEs and synergize cancer immunotherapy with checkpoint therapies, with minimized systemic toxicity.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Imunoterapia/métodos , Nanogéis/administração & dosagem , Neoplasias/tratamento farmacológico , Animais , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Composição de Medicamentos/métodos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Injeções Intralesionais , Lipossomos , Camundongos , Nanogéis/química , Recidiva Local de Neoplasia/prevenção & controle , Neoplasias/imunologia , Neoplasias/patologia , Seringas , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
8.
Adv Mater ; 31(34): e1803322, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30773696

RESUMO

Cancer immunotherapies that harness the body's immune system to combat tumors have received extensive attention and become mainstream strategies for treating cancer. Despite promising results, some problems remain, such as the limited patient response rate and the emergence of severe immune-related adverse effects. For most patients, the therapeutic efficacy of cancer immunotherapy is mainly limited by the immunosuppressive tumor microenvironment (TME). To overcome such obstacles in the TME, the immunomodulation of immunosuppressive factors and therapeutic immune cells (e.g., T cells and antigen-presenting cells) should be carefully designed and evaluated. Nanoengineered synthetic immune niches have emerged as highly customizable platforms with a potent capability for reprogramming the immunosuppressive TME. Here, recent developments in nano-biomaterials that are rationally designed to modulate the immunosuppressive TME in a spatiotemporal manner for enhanced cancer immunotherapy which are rationally designed to modulate the immunosuppressive TME in a spatiotemporal manner for enhanced cancer immunotherapy are highlighted.


Assuntos
Imunomodulação , Nanoestruturas/química , Neoplasias/terapia , Microambiente Tumoral/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Sistemas de Liberação de Medicamentos , Epigênese Genética , Humanos , Terapia de Imunossupressão , Imunoterapia , Terapia de Alvo Molecular , Neoplasias/imunologia , Linfócitos T/imunologia
9.
Adv Mater ; 30(18): e1706719, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29572968

RESUMO

The development of biomaterial-based immune niches that can modulate immunosuppressive factors in tumor microenvironment (TME) will be a key technology for improving current cancer immunotherapy. Here, implantable, engineered 3D porous scaffolds are designed to generate synergistic action between myeloid-derived suppressor cell (MDSC)-depleting agents, which can accommodate the establishment of a permissive immunogenic microenvironment to counteract tumor-induced immunosuppression, and cancer vaccines consisting of whole tumor lysates and nanogel-based adjuvants, which can generate tumor antigen-specific T cell responses. The local peritumoral implantation of the synthetic immune niche (termed immuneCare-DISC, iCD) as a postsurgical treatment in an advanced-stage primary 4T1 breast tumor model generates systemic antitumor immunity and prevents tumor recurrence at the surgical site as well as the migration of residual tumor cells into the lungs, resulting in 100% survival. These therapeutic outcomes are achieved through the inhibition of immunosuppressive MDSCs in tumors and spleens by releasing gemcitabine and recruitment/activation of dendritic cells, enhanced population of CD4+ and CD8+ T cells, and increased IFN-γ production by cancer vaccines from the iCD. This combined spatiotemporal modulation of tumor-derived immunosuppression and vaccine-induced immune stimulation through the iCD is expected to provide an immune niche for prevention of postoperative tumor recurrence and metastasis.


Assuntos
Imunoterapia , Linfócitos T CD8-Positivos , Vacinas Anticâncer , Humanos , Terapia de Imunossupressão , Microambiente Tumoral
10.
Int J Nanomedicine ; 12: 7501-7517, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29066896

RESUMO

In this study, we suggest a designer vaccine adjuvant that can mimic the drainage of pathogens into lymph nodes and activate innate immune response in lymph nodes. By the amination of multivalent carboxyl groups in poly-(γ-glutamic acid) (γ-PGA) nanomicelles, the size was reduced for rapid entry into lymphatic vessels, and the immunologically inert nanomicelles were turned into potential activators of inflammasomes. Aminated γ-PGA nanomicelles (aPNMs) induced NLRP3 inflammasome activation and the subsequent release of proinflammatory IL-1ß. The NLRP3-dependent inflammasome induction mechanism was confirmed through enzyme (cathepsin B and caspase-1) inhibitors and NLRP3 knockout mice model. After the aPNMs were combined with a clinically evaluated TLR3 agonist, polyinosinic-polycytidylic acid sodium salt (aPNM-IC), they triggered multiple arms of the innate immune response, including the secretion of pro-inflammatory cytokines by both inflammasomes and an inflammasome-independent pathway and the included type I interferons.


Assuntos
Adjuvantes Imunológicos/farmacologia , Aminas/química , Imunidade Inata , Inflamassomos/metabolismo , Linfonodos/imunologia , Micelas , Nanopartículas/química , Vacinas/imunologia , Animais , Caspase 1/metabolismo , Feminino , Imunidade Inata/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Poli I-C/farmacologia , Ácido Poliglutâmico/análogos & derivados , Ácido Poliglutâmico/química
11.
Pharm Dev Technol ; 21(1): 8-13, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25220889

RESUMO

Chlorophene-loaded nanospheres with various formulation parameters were evaluated. The optimal formulation was found at 0.1% w/v of poloxamer 407, 15 mL of ethyl acetate and 20% initial chlorophene loading that provided the suitable size (179 nm), the highest loading content (19.2%), encapsulation efficiency (88.0%) and yield (91.6%). Moreover, encapsulation of chlorophene in nanospheres was able to prolong and sustain drug release over one month. Chlorophene-loaded nanospheres were effective against Staphylococcus aureus (S. aureus) and Candida albicans (C. albicans), the main cause of hospital-acquired infections. Chlorophene-loaded nanospheres were effective against S. aureus (>46 µg/mL) and C. albicans (>184 µg/mL). These nanospheres appeared to have profound effect on the time-dependent hemolytic activity due to gradual release of chlorophene. At the concentration of 46 µg/mL, nearly no HRBC hemolysis in 24 h compared to 80% of hemolysis from free drug. In conclusion, polymeric nanospheres were successfully fabricated to encapsulate chlorophene which can eliminate inherent toxicity of drugs and have potential uses in prolonged release of antimicrobial.


Assuntos
Anti-Infecciosos/síntese química , Diclorofeno/análogos & derivados , Sistemas de Liberação de Medicamentos/métodos , Nanosferas/química , Anti-Infecciosos/administração & dosagem , Candida albicans/efeitos dos fármacos , Candida albicans/fisiologia , Química Farmacêutica , Preparações de Ação Retardada , Diclorofeno/administração & dosagem , Diclorofeno/síntese química , Relação Dose-Resposta a Droga , Humanos , Testes de Sensibilidade Microbiana/métodos , Nanosferas/administração & dosagem , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia
12.
J Mater Sci Mater Med ; 26(2): 78, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25631275

RESUMO

Urinary tract infections (UTIs) are the most common type of hospital-acquired infection which cause significant morbidity and mortality. Antibacterial urinary devices to prevent UTIs are in great demand, while the problem of releasing antibacterials is still limited by duration of antibacterial release and hinders their clinical applications. This study investigated a new approach to sustain release of chlorhexidine (CHX) from urinary devices by coating of chlorhexidine-loaded nanospheres (CHX-NPs) on the surface. CHX-NPs were prepared by high-pressure emulsification-solvent evaporation technique that provided the size of nanospheres at 198.8 nm and the drug loading content at 5.6%. These nanospheres were spray-coated on silicone surface with reproducible and predictable amount of CHX. Release studies conducted in artificial urine to mimic in vivo condition showed that suitable dose of CHX was released in a sustained manner within a couple of weeks. Additionally, CHX-NPs showed antibacterial activity against common bacteria causing UTIs up to 15 days, which is threefold longer than that of physical mixing between CHX and polymer. Results from this study suggest possible applications of CHX-NPs in coating the surface of ureteral-relating devices for sustained antibacterial release.


Assuntos
Fenômenos Fisiológicos Bacterianos/efeitos dos fármacos , Clorexidina/administração & dosagem , Preparações de Ação Retardada/administração & dosagem , Nanosferas/química , Silicones/química , Cateteres Urinários/microbiologia , Adsorção , Antibacterianos/administração & dosagem , Antibacterianos/química , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Clorexidina/química , Materiais Revestidos Biocompatíveis/administração & dosagem , Materiais Revestidos Biocompatíveis/síntese química , Preparações de Ação Retardada/síntese química , Difusão , Nanocápsulas/administração & dosagem , Nanocápsulas/química , Nanocápsulas/ultraestrutura , Nanosferas/administração & dosagem , Nanosferas/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...